1
|
Regenold M, Bannigan P, Evans JC, Waspe A, Temple MJ, Allen C. Turning down the heat: The case for mild hyperthermia and thermosensitive liposomes. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2022; 40:102484. [PMID: 34748961 DOI: 10.1016/j.nano.2021.102484] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/09/2021] [Accepted: 09/30/2021] [Indexed: 12/17/2022]
Abstract
"A single disappointing study does not mean an end to the future of ThermoDox®", writes Michael Tardugno (CEO of Celsion Corporation), after announcing the termination of Celsion's second Phase III clinical trial. The OPTIMA trial, as it was known, evaluated their thermosensitive liposome (TSL) formulation of doxorubicin (ThermoDox®) in combination with radiofrequency ablation for the treatment of hepatocellular carcinoma (HCC). The purpose of this perspective is to review the case of ThermoDox and to address questions related to its clinical translation. Specifically, what has prevented the clinical translation of this once highly regarded breakthrough technology? Is this the end of TSLs? What can we learn from the challenges faced in the clinical development of this multi-modal therapy? As formulation scientists working in the field, we continue to believe that heat-triggered drug delivery platforms have tremendous potential as chemotherapy. Herein, we highlight potential limitations in the design of many of the Thermodox clinical trials, and we propose that despite these setbacks, TSLs have the potential to become an effective component of cancer therapy.
Collapse
Affiliation(s)
- Maximilian Regenold
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Pauric Bannigan
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - James C Evans
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Adam Waspe
- Department of Medical Imaging, University of Toronto, Toronto, Ontario, Canada; Centre for Image-Guided Innovation and Therapeutic Intervention, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Michael J Temple
- Department of Medical Imaging, University of Toronto, Toronto, Ontario, Canada; Image Guided Therapy, Diagnostic Imaging, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Christine Allen
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
2
|
Sang R, Stratton B, Engel A, Deng W. Liposome technologies towards colorectal cancer therapeutics. Acta Biomater 2021; 127:24-40. [PMID: 33812076 DOI: 10.1016/j.actbio.2021.03.055] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 03/05/2021] [Accepted: 03/23/2021] [Indexed: 02/08/2023]
Abstract
Colorectal cancer (CRC) is the third most common cancer and the fourth most common deadly cancer worldwide. After treatment with curative intent recurrence rates vary with staging 0-13% in Stage 1, 11-61% in S2 and 28-73% in Stage 3. The toxicity to healthy tissues from chemotherapy and radiotherapy and drug resistance severely affect the quality of life and cancer specific outcomes of CRC patients. To overcome some of these limitations, many efforts have been made to develop nanomaterial-based drug delivery systems. Among these nanocarriers, liposomes represented one of the most successful candidates in delivering targeted oncological treatment, improving safety profile and therapeutic efficacy of encapsulated drugs. In this review we will discuss liposome design with a particular focus on the targeting feature and triggering functions. We will also summarise the recent advances in liposomal delivery system for CRC treatment in both the preclinical and clinical studies. We will finally provide our perspectives on the liposome technology development for the future clinical translation. STATEMENT OF SIGNIFICANCE: Conventional treatments for colorectal cancer (CRC) severely affect the therapeutic effects for advanced patients. With the development of nanomedicines, liposomal delivery system appears to be one of the most promising nanocarriers for CRC treatment. In last three years several reviews in this area have been published focusing on the preclinical research and drug delivery function, which is a fairly narrow focus in the field of liposome technology for CRC therapy. Our review presented the most recent advances of the liposome technology (both clinical and preclinical applications) for CRC with strong potential for further clinical translation. We believe it will attract lots of attention from various audiences, including researchers, clinicians and the industry.
Collapse
|
3
|
Mechanistic investigation of thermosensitive liposome immunogenicity and understanding the drivers for circulation half-life: A polyethylene glycol versus 1,2-dipalmitoyl-sn-glycero-3-phosphodiglycerol study. J Control Release 2021; 333:1-15. [DOI: 10.1016/j.jconrel.2021.03.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 03/10/2021] [Indexed: 12/18/2022]
|
4
|
Monahan DS, Almas T, Wyile R, Cheema FH, Duffy GP, Hameed A. Towards the use of localised delivery strategies to counteract cancer therapy-induced cardiotoxicities. Drug Deliv Transl Res 2021; 11:1924-1942. [PMID: 33449342 DOI: 10.1007/s13346-020-00885-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2020] [Indexed: 02/06/2023]
Abstract
Cancer therapies have significantly improved cancer survival; however, these therapies can often result in undesired side effects to off target organs. Cardiac disease ranging from mild hypertension to heart failure can occur as a result of cancer therapies. This can warrant the discontinuation of cancer treatment in patients which can be detrimental, especially when the treatment is effective. There is an urgent need to mitigate cardiac disease that occurs as a result of cancer therapy. Delivery strategies such as the use of nanoparticles, hydrogels, and medical devices can be used to localise the treatment to the tumour and prevent off target side effects. This review summarises the advancements in localised delivery of anti-cancer therapies to tumours. It also examines the localised delivery of cardioprotectants to the heart for patients with systemic disease such as leukaemia where localised tumour delivery might not be an option.
Collapse
Affiliation(s)
- David S Monahan
- Anatomy & Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine Nursing and Health Science, National University of Ireland Galway, Galway, Ireland.,Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Galway, Ireland.,Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Talal Almas
- School of Medicine, RCSI University of Medicine and Health Sciences, 123, St. Stephens Green, Dublin 2, Dublin, D02 YN77, Ireland
| | - Robert Wyile
- Anatomy & Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine Nursing and Health Science, National University of Ireland Galway, Galway, Ireland
| | - Faisal H Cheema
- HCA Healthcare, Gulf Coast Division, Houston, TX, USA.,College of Medicine, University of Houston, Houston, TX, USA
| | - Garry P Duffy
- Anatomy & Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine Nursing and Health Science, National University of Ireland Galway, Galway, Ireland.,Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Galway, Ireland.,Tissue Engineering Research Group (TERG), Department of Anatomy, RCSI University of Medicine and Health Sciences, 123, St. Stephens Green, Dublin 2, Dublin, D02 YN77, Ireland.,Advanced Materials for Biomedical Engineering and Regenerative Medicine (AMBER), National University of Ireland, Trinity College Dublin &, Galway, Ireland.,Trinity Centre for Biomedical Engineering (TCBE), Trinity College Dublin, Dublin 2, Dublin, Ireland
| | - Aamir Hameed
- Tissue Engineering Research Group (TERG), Department of Anatomy, RCSI University of Medicine and Health Sciences, 123, St. Stephens Green, Dublin 2, Dublin, D02 YN77, Ireland. .,Trinity Centre for Biomedical Engineering (TCBE), Trinity College Dublin, Dublin 2, Dublin, Ireland.
| |
Collapse
|
5
|
Amrahli M, Centelles M, Cressey P, Prusevicius M, Gedroyc W, Xu XY, So PW, Wright M, Thanou M. MR-labelled liposomes and focused ultrasound for spatiotemporally controlled drug release in triple negative breast cancers in mice. Nanotheranostics 2021; 5:125-142. [PMID: 33457192 PMCID: PMC7806456 DOI: 10.7150/ntno.52168] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 11/30/2020] [Indexed: 01/12/2023] Open
Abstract
Rationale: Image-guided, triggerable, drug delivery systems allow for precisely placed and highly localised anti-cancer treatment. They contain labels for spatial mapping and tissue uptake tracking, providing key location and timing information for the application of an external stimulus to trigger drug release. High Intensity Focused Ultrasound (HIFU or FUS) is a non-invasive approach for treating small tissue volumes and is particularly effective at inducing drug release from thermosensitive nanocarriers. Here, we present a novel MR-imageable thermosensitive liposome (iTSL) for drug delivery to triple-negative breast cancers (TNBC). Methods: A macrocyclic gadolinium-based Magnetic Resonance Imaging (MRI) contrast agent was covalently linked to a lipid. This was incorporated at 30 mol% into the lipid bilayer of a thermosensitive liposome that was also encapsulating doxorubicin. The resulting iTSL-DOX formulation was assessed for physical and chemical properties, storage stability, leakage of gadolinium or doxorubicin, and thermal- or FUS-induced drug release. Its effect on MRI relaxation time was tested in phantoms. Mice with tumours were used for studies to assess both tumour distribution and contrast enhancement over time. A lipid-conjugated near-infrared fluorescence (NIRF) probe was also included in the liposome to facilitate the real time monitoring of iTSL distribution and drug release in tumours by NIRF bioimaging. TNBC (MDA-MB-231) tumour-bearing mice were then used to demonstrate the efficacy at retarding tumour growth and increasing survival. Results: iTSL-DOX provided rapid FUS-induced drug release that was dependent on the acoustic power applied. It was otherwise found to be stable, with minimum leakage of drug and gadolinium into buffers or under challenging conditions. In contrast to the usually suggested longer FUS treatment we identified that brief (~3 min) FUS significantly enhanced iTSL-DOX uptake to a targeted tumour and triggered near-total release of encapsulated doxorubicin, causing significant growth inhibition in the TNBC mouse model. A distinct reduction in the tumours' average T1 relaxation times was attributed to the iTSL accumulation. Conclusions: We demonstrate that tracking iTSL in tumours using MRI assists the application of FUS for precise drug release and therapy.
Collapse
Affiliation(s)
- Maral Amrahli
- School of Cancer & Pharmaceutical Sciences, King's College London, U.K
| | - Miguel Centelles
- School of Cancer & Pharmaceutical Sciences, King's College London, U.K
| | - Paul Cressey
- School of Cancer & Pharmaceutical Sciences, King's College London, U.K
| | | | | | - Xiao Yun Xu
- Department of Chemical Engineering, Imperial College London, U.K
| | - Po-Wah So
- Department of Neuroimaging, King's College London, U.K
| | - Michael Wright
- School of Cancer & Pharmaceutical Sciences, King's College London, U.K
| | - Maya Thanou
- School of Cancer & Pharmaceutical Sciences, King's College London, U.K
| |
Collapse
|
6
|
Datta NR, Kok HP, Crezee H, Gaipl US, Bodis S. Integrating Loco-Regional Hyperthermia Into the Current Oncology Practice: SWOT and TOWS Analyses. Front Oncol 2020; 10:819. [PMID: 32596144 PMCID: PMC7303270 DOI: 10.3389/fonc.2020.00819] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 04/27/2020] [Indexed: 12/14/2022] Open
Abstract
Moderate hyperthermia at temperatures between 40 and 44°C is a multifaceted therapeutic modality. It is a potent radiosensitizer, interacts favorably with a host of chemotherapeutic agents, and, in combination with radiotherapy, enforces immunomodulation akin to “in situ tumor vaccination.” By sensitizing hypoxic tumor cells and inhibiting repair of radiotherapy-induced DNA damage, the properties of hyperthermia delivered together with photons might provide a tumor-selective therapeutic advantage analogous to high linear energy transfer (LET) neutrons, but with less normal tissue toxicity. Furthermore, the high LET attributes of hyperthermia thermoradiobiologically are likely to enhance low LET protons; thus, proton thermoradiotherapy would mimic 12C ion therapy. Hyperthermia with radiotherapy and/or chemotherapy substantially improves therapeutic outcomes without enhancing normal tissue morbidities, yielding level I evidence reported in several randomized clinical trials, systematic reviews, and meta-analyses for various tumor sites. Technological advancements in hyperthermia delivery, advancements in hyperthermia treatment planning, online invasive and non-invasive MR-guided thermometry, and adherence to quality assurance guidelines have ensured safe and effective delivery of hyperthermia to the target region. Novel biological modeling permits integration of hyperthermia and radiotherapy treatment plans. Further, hyperthermia along with immune checkpoint inhibitors and DNA damage repair inhibitors could further augment the therapeutic efficacy resulting in synthetic lethality. Additionally, hyperthermia induced by magnetic nanoparticles coupled to selective payloads, namely, tumor-specific radiotheranostics (for both tumor imaging and radionuclide therapy), chemotherapeutic drugs, immunotherapeutic agents, and gene silencing, could provide a comprehensive tumor-specific theranostic modality akin to “magic (nano)bullets.” To get a realistic overview of the strength (S), weakness (W), opportunities (O), and threats (T) of hyperthermia, a SWOT analysis has been undertaken. Additionally, a TOWS analysis categorizes future strategies to facilitate further integration of hyperthermia with the current treatment modalities. These could gainfully accomplish a safe, versatile, and cost-effective enhancement of the existing therapeutic armamentarium to improve outcomes in clinical oncology.
Collapse
Affiliation(s)
- Niloy R Datta
- Centre for Radiation Oncology KSA-KSB, Kantonsspital Aarau, Aarau, Switzerland
| | - H Petra Kok
- Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Hans Crezee
- Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Udo S Gaipl
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Stephan Bodis
- Centre for Radiation Oncology KSA-KSB, Kantonsspital Aarau, Aarau, Switzerland
| |
Collapse
|
7
|
Chi X, Liu K, Luo X, Yin Z, Lin H, Gao J. Recent advances of nanomedicines for liver cancer therapy. J Mater Chem B 2020; 8:3747-3771. [DOI: 10.1039/c9tb02871d] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
This review highlights recent advancements in nanomedicines for liver cancer therapy.
Collapse
Affiliation(s)
- Xiaoqin Chi
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma
- Zhongshan Hospital
- Xiamen University
- Xiamen 361004
- China
| | - Kun Liu
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation
- The Key Laboratory for Chemical Biology of Fujian Province, and Department of Chemical Biology
- College of Chemistry and Chemical Engineering
- Xiamen University
- Xiamen 361005
| | - Xiangjie Luo
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation
- The Key Laboratory for Chemical Biology of Fujian Province, and Department of Chemical Biology
- College of Chemistry and Chemical Engineering
- Xiamen University
- Xiamen 361005
| | - Zhenyu Yin
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma
- Zhongshan Hospital
- Xiamen University
- Xiamen 361004
- China
| | - Hongyu Lin
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation
- The Key Laboratory for Chemical Biology of Fujian Province, and Department of Chemical Biology
- College of Chemistry and Chemical Engineering
- Xiamen University
- Xiamen 361005
| | - Jinhao Gao
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation
- The Key Laboratory for Chemical Biology of Fujian Province, and Department of Chemical Biology
- College of Chemistry and Chemical Engineering
- Xiamen University
- Xiamen 361005
| |
Collapse
|
8
|
Abstract
Liposomes have been employed as cancer therapy clinically since the 1990s, with the primary benefit of reduced toxicity but no appreciable efficacy improvement. Thermosensitive liposomes (TSLs) are specifically formulated such that they release the encapsulated drug when exposed to hyperthermic temperatures in the fever range (~40-42°C) and have been investigated as cancer therapy for several decades, with first clinical trials initiated in the last decade. Combined with localized hyperthermia, TSLs allow precise drug delivery to a targeted region. Typically, the targeted tissue is exposed to localized hyperthermia facilitated by an image-guided hyperthermia device. Thus, TSLs enable image-guided drug delivery where drug is delivered to a tissue region identified by medical imaging. Recent TSL formulations are based on the more recent paradigm of intravascular triggered release, where drug is released rapidly (within seconds) while TSLs pass through the vasculature of the heated tissue region. The drug released within the blood then extravasates and is taken up by cancer cells. These TSLs enable up to 20-30 times higher tumor drug uptake compared to infusion of unencapsulated drug, and the dose locally delivered to the heated region can be modulated based on heating duration. This chapter reviews various TSL formulations, the different anticancer agents that have been encapsulated, as well as targeted cancer types. Further, the various hyperthermia devices that have been used for image-guided hyperthermia are reviewed, focusing on those that have been employed in human patients.
Collapse
Affiliation(s)
- Dieter Haemmerich
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, United States.
| | - Anjan Motamarry
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, United States; Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
9
|
Bi H, Xue J, Jiang H, Gao S, Yang D, Fang Y, Shi K. Current developments in drug delivery with thermosensitive liposomes. Asian J Pharm Sci 2019; 14:365-379. [PMID: 32104466 PMCID: PMC7032122 DOI: 10.1016/j.ajps.2018.07.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 06/26/2018] [Accepted: 07/25/2018] [Indexed: 12/19/2022] Open
Abstract
Thermosensitive liposomes (TSLs) have been an important research area in the field of tumor targeted chemotherapy. Since the first TSLs appeared that using 1,2-dipalmitoyl-sn-glyce-ro-3-phosphocholine (DPPC) as the primary liposomal lipid, many studies have been done using this type of liposome from basic and practical aspects. While TSLs composed of DPPC enhance the cargo release near the phase transition temperature, it has been shown that many factors affect their temperature sensitivity. Thus numerous attempts have been undertaken to develop new TSLs for improving their thermal response performance. The main objective of this review is to introduce the development and recent update of innovative TSLs formulations, including combination of radiofrequency ablation (RFA), high-intensity focused ultrasound (HIFU), magnetic resonance imaging (MRI) and alternating magnetic field (AMF). In addition, various factors affecting the design of TSLs, such as lipid composition, surfactant, size and serum components are also discussed.
Collapse
Key Words
- (DPPC), 1,2-dipalmitoyl-sn-glycero-3-phosphatidylcholine
- (DPPGOG), 1,2-dipalmitoyl-sn-glycero-3-phosphoglyceroglycerol
- (DSPC), 1,2-distearoyl-sn-glycero-3-phosphocholine
- (DSPE-mPEG2000), 1,2-distearoyl-sn-glycero-3-phosphatiylethanol-amine-N-[methoxy(polyethyleneglycol)-2000]
- (LTSLs), lyso-lipid temperature sensitive liposomes
- (MPPC), 1-myristoyl-2-palmitoyl-sn-glycero-3-phosphatidylcholine
- (MSPC), 1-stearoyl-2-hydroxy-sn-glycero-3-phosphatidylcholine
- (P-lyso-PC), lysophosphatidylcholine
- (P188), 1-palmitoyl-2-stearoyl-sn-glycero-3-phosphatidylcholinex
- (P188), HO-(C2H4O)a-(C3H6O)b-(C2H4O)c-H, a=80, b=27, c=80
- Content release rate
- Drug delivery
- Hyperthermia
- Smart liposomes
- Thermosensitive liposomes
- Tumor chemotherapy
- fTSLs, fast release TSLs
- sTSLs, slow release TSLs
Collapse
Affiliation(s)
- Hongshu Bi
- Institute of New Drug Development, Liaoning Yaolian Pharmaceutical Co., Ltd., Benxi, Liaoning 117004, China
| | - Jianxiu Xue
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, Liaoning 117004, China
| | - Hong Jiang
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, Liaoning 117004, China
| | - Shan Gao
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, Liaoning 117004, China
| | - Dongjuan Yang
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, Liaoning 117004, China
| | - Yan Fang
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, Liaoning 117004, China
| | - Kai Shi
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, Liaoning 117004, China
| |
Collapse
|
10
|
Enhanced Drug Delivery to the Skin Using Liposomes. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2018; 6:e1739. [PMID: 30175003 PMCID: PMC6110675 DOI: 10.1097/gox.0000000000001739] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 02/07/2018] [Indexed: 12/02/2022]
Abstract
Background: Enhancing drug delivery to the skin has importance in many therapeutic strategies. In particular, the outcome in vascularized composite allotransplantation mainly depends on systemic immunosuppression to prevent and treat episodes of transplant rejection. However, the side effects of systemic immunosuppression may introduce substantial risk to the patient and are weighed against the expected benefits. Successful enhancement of delivery of immunosuppressive agents to the most immunogenic tissues would allow for a reduction in systemic doses, thereby minimizing side effects. Nanoparticle-assisted transport by low temperature–sensitive liposomes (LTSLs) has shown some benefit in anticancer therapy. Our goal was to test whether delivery of a marker agent to the skin could be selectively enhanced. Methods: In an in vivo model, LTSLs containing doxorubicin (dox) as a marker were administered intravenously to rats that were exposed locally to mild hyperthermia. Skin samples of the hyperthermia treated hind limb were compared with skin of the contralateral normothermia hind limb. Tissue content of dox was quantified both via high-performance liquid chromatography and via histology in skin and liver. Results: The concentration of dox in hyperthermia-treated skin was significantly elevated over both normothermic skin and liver. (P < 0.02). Conclusions: We show here that delivery of therapeutics to the skin can be targeted and enhanced using LTSLs. Targeting drug delivery with this method may reduce the systemic toxicity seen in a systemic free-drug administration. Development of more hydrophilic immunosuppressants in the future would increase the applicability of this system in the treatment of rejection reactions in vascularized composite allotransplantation. The treatment of other skin condition might be another potential application.
Collapse
|
11
|
Fu X, Lu Y, Guo J, Liu H, Deng A, Kuang C, Xie X. K237-modified thermosensitive liposome enhanced the delivery efficiency and cytotoxicity of paclitaxel in vitro. J Liposome Res 2018; 29:86-93. [PMID: 29671386 DOI: 10.1080/08982104.2018.1458863] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
This study aimed to develop novel temperature-sensitive liposomes loading paclitaxel (PTX-TSL) and evaluate them in vitro to improve the delivery efficiency and targeting of PTX. K237 peptide was conjugated to the terminal NHS of 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[hydroxyl succinimidyl (polyethylene glycol)-(DSPE-PEG-NHS), and K237-modified PTX-TSL (K237-PTX-TSL) was prepared using a film dispersion method. K237-TSL encapsulation with calcein was synthesized and used to determine the cellular uptake of TSL. The morphology of K237-PTX-TSL was observed using a transmission electron microscope. The particle size and potential were measured using a laser particle size analyzer. The phase transition temperature was detected using the differential scanning calorimetry. The Cell Counting Kit-8 assay and flow cytometry were used to evaluate the effects of K237-PTX-TSL on the proliferation and cell cycle of cell lines SKOV-3 and human umbilical vein endothelial cell (HUVEC). The encapsulation efficiency of K237-PTX-TSL was 94.23% ± 0.76%. The particle diameter was 88.3 ± 4.7 nm. K237-PTX-TSL showed a fast release profile at 42 °C, while it was stable at 37 °C. PTX-TSL combined with hyperthermia significantly inhibited the cell proliferation of SKOV-3 cells and HUVECs due to increased cell arrest in the G2/M phase. The half-minimal inhibitory concentration value of K237-PTX-TSL on SKOV-3 cells and HUVECs was 13.61 ± 1.81 and 5.54 ± 0.95 nmol/L, respectively, which were significantly lower than those with PTX-TSL (p < 0.01). K237 modification could increase the targeting efficiency of TSL to cancer cells and vascular endothelial cells, thus resulting in higher cytotoxicities compared with PTX-TSL, which might be a potential formulation for targeting cancer therapy.
Collapse
Affiliation(s)
- Xudong Fu
- a Department of Pharmacy , Wuhan General Hospital of Chinese PLA , Wuhan , China
| | - Yuanyuan Lu
- a Department of Pharmacy , Wuhan General Hospital of Chinese PLA , Wuhan , China.,b Department of Pharmacy , The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Jiaping Guo
- c Department of Maxillofacial Surgery , Wuhan General Hospital of Chinese PLA , Wuhan , China
| | - Hui Liu
- a Department of Pharmacy , Wuhan General Hospital of Chinese PLA , Wuhan , China
| | - Aiping Deng
- b Department of Pharmacy , The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Changchun Kuang
- a Department of Pharmacy , Wuhan General Hospital of Chinese PLA , Wuhan , China
| | - Xiangyang Xie
- a Department of Pharmacy , Wuhan General Hospital of Chinese PLA , Wuhan , China
| |
Collapse
|
12
|
Formation of protein corona in vivo affects drug release from temperature-sensitive liposomes. J Control Release 2018. [DOI: 10.1016/j.jconrel.2018.02.038] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
13
|
Chitgupi U, Shao S, Carter KA, Huang WC, Lovell JF. Multicolor Liposome Mixtures for Selective and Selectable Cargo Release. NANO LETTERS 2018; 18:1331-1336. [PMID: 29384679 DOI: 10.1021/acs.nanolett.7b05025] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Many approaches exist for stimuli-triggered cargo release from nanocarriers, but few can provide for on-demand release of multiple payloads, selectively. Here, we report the synthesis of purpurin-phospholipid (Pur-P), a lipid chromophore that has near-infrared absorbance red-shifted by 30 nm compared to a structurally similar pyropheophorbide-phospholipid (Pyr-P). Liposomes containing small amounts of either Pur-P or Pyr-P exhibited similar physical properties and fluorescence self-quenching. Loaded with distinct cargos, Pur-P and Pyr-P liposomes were mixed into a single colloidal suspension and selectively released cargo depending on irradiation wavelength. Spatiotemporal control of distinct cargo release was achieved by controlling multicolor laser placement. Using basic orange and doxorubicin anthraquinones, multidimensional cytotoxicity gradients were established to gauge efficacy against cancer cells using light-released drug. Wavelength selectivity of cargo release was maintained following intramuscular administration to mice.
Collapse
Affiliation(s)
- Upendra Chitgupi
- Department of Biomedical Engineering, University at Buffalo, State University of New York , Buffalo, New York 14260, United States
| | - Shuai Shao
- Department of Biomedical Engineering, University at Buffalo, State University of New York , Buffalo, New York 14260, United States
| | - Kevin A Carter
- Department of Biomedical Engineering, University at Buffalo, State University of New York , Buffalo, New York 14260, United States
| | - Wei-Chiao Huang
- Department of Biomedical Engineering, University at Buffalo, State University of New York , Buffalo, New York 14260, United States
| | - Jonathan F Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York , Buffalo, New York 14260, United States
| |
Collapse
|
14
|
Rehman M, Madni A, Shi D, Ihsan A, Tahir N, Chang KR, Javed I, Webster TJ. Enhanced blood brain barrier permeability and glioblastoma cell targeting via thermoresponsive lipid nanoparticles. NANOSCALE 2017; 9:15434-15440. [PMID: 28976512 DOI: 10.1039/c7nr05216b] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Thermoresponsive targeting is used to deliver therapeutic agents at hyperthermic conditions (39-45 °C). However, available thermoresponsive drug delivery systems (TDDS), including liposomes, have a complex method of preparation involving toxic solvents and reagents. The objective of this in vitro study was to prepare and characterize thermoresponsive lipid nanoparticles (TLN) for treating glioblastoma, the most aggressive brain tumor whose treatment is limited by a low blood brain barrier (BBB) permeability of drugs. Thermoresponsive lipids were prepared by mixing liquid and solid fatty acids (0.1 : 1 to 2 : 1 ratio) and lipid mixtures exhibiting a solid-liquid phase transition at 39 °C were identified by plotting melting point against liquid contents. TLN were prepared by a hot melt encapsulation method using mono- or double-surfactant systems. TLN showed desirable size (<270 nm), zeta potential (-35 to -50 mV), spherical morphology and stability by FTIR studies. In the drug release studies, paclitaxel release was slow at 37 °C, however, it was released abruptly at 39 °C due to the faster diffusion rate from liquid state nanoparticles. During cytotoxicity studies, the unloaded TLN were non-toxic whereas paclitaxel loaded TLN showed higher cytotoxicity to glioblastoma cells at 39 °C (69% cell viability after one hour) compared to 37 °C (82% cell viability). The TLN showed higher permeability across an in vitro model of BBB at 39 °C due to a deformable liquid state which can squeeze through the tight junctions of the BBB. In conclusion, this study demonstrated that the TLN can be used as a safe and effective alternative to traditional TDDS with higher potential to target glioblastoma cells across the BBB.
Collapse
Affiliation(s)
- M Rehman
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA.
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Mertz D, Sandre O, Bégin-Colin S. Drug releasing nanoplatforms activated by alternating magnetic fields. Biochim Biophys Acta Gen Subj 2017; 1861:1617-1641. [PMID: 28238734 DOI: 10.1016/j.bbagen.2017.02.025] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 02/17/2017] [Accepted: 02/20/2017] [Indexed: 02/05/2023]
Abstract
The use of an alternating magnetic field (AMF) to generate non-invasively and spatially a localized heating from a magnetic nano-mediator has become very popular these last years to develop magnetic hyperthermia (MH) as a promising therapeutic modality already used in the clinics. AMF has become highly attractive this last decade over others radiations, as AMF allows a deeper penetration in the body and a less harmful ionizing effect. In addition to pure MH which induces tumor cell death through local T elevation, this AMF-generated magneto-thermal effect can also be exploited as a relevant external stimulus to trigger a drug release from drug-loaded magnetic nanocarriers, temporally and spatially. This review article is focused especially on this concept of AMF induced drug release, possibly combined with MH. The design of such magnetically responsive drug delivery nanoplatforms requires two key and complementary components: a magnetic mediator which collects and turns the magnetic energy into local heat, and a thermoresponsive carrier ensuring thermo-induced drug release, as a consequence of magnetic stimulus. A wide panel of magnetic nanomaterials/chemistries and processes are currently developed to achieve such nanoplatforms. This review article presents a broad overview about the fundamental concepts of drug releasing nanoplatforms activated by AMF, their formulations, and their efficiency in vitro and in vivo. This article is part of a Special Issue entitled "Recent Advances in Bionanomaterials" Guest Editors: Dr. Marie-Louise Saboungi and Dr. Samuel D. Bader.
Collapse
Affiliation(s)
- Damien Mertz
- Institut de Physique et Chimie des Matériaux de Strasbourg, UMR 7504 CNRS, Université de Strasbourg, 23, rue du Loess, 67034 Strasbourg, France.
| | - Olivier Sandre
- Laboratoire de Chimie des Polymères Organiques (LCPO), CNRS UMR 5629, Université de Bordeaux, Bordeaux-INP, Pessac 33607, Cedex, France
| | - Sylvie Bégin-Colin
- Institut de Physique et Chimie des Matériaux de Strasbourg, UMR 7504 CNRS, Université de Strasbourg, 23, rue du Loess, 67034 Strasbourg, France
| |
Collapse
|
16
|
Abstract
Hepatocellular carcinoma (HCC), also called malignant hepatoma, is one of the deadliest cancers due to its complexities, reoccurrence after surgical resection, metastasis and heterogeneity. Incidence and mortality of HCC are increasing in Western countries and are expected to rise as a consequence of the obesity epidemic. Multiple factors trigger the initiation and progression of HCC including chronic alcohol consumption, viral hepatitis B and C infection, metabolic disorders and age. Although Sorafenib is the only FDA approved drug for the treatment of HCC, numerous treatment modalities such as transcatheter arterial chemoembolization/transarterial chemoembolization (TACE), radiotherapy, locoregional therapy and chemotherapy have been tested in the clinics. Polymeric nanoparticles, liposomes, and micelles carrying small molecules, proteins, peptides and nucleic acids have attracted great attention for the treatment of various cancers including HCC. Herein, we discuss the pathogenesis of HCC in relation to its various recent treatment methodologies using nanodelivery of monoclonal antibodies (mAbs), small molecules, miRNAs and peptides. Synopsis of recent clinical trials of mAbs and peptide drugs has been presented with a broad overview of the pathogenesis of the disease and treatment efficacy.
Collapse
Affiliation(s)
- Rinku Dutta
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Ram I Mahato
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States.
| |
Collapse
|
17
|
Tang HX, Zhao TW, Zheng T, Sheng YJ, Zheng HS, Zhang YS. Liver-targeting liposome drug delivery system and its research progress in liver diseases. Shijie Huaren Xiaohua Zazhi 2016; 24:4238-4246. [DOI: 10.11569/wcjd.v24.i31.4238] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Liposome-based targeted therapy is mainly divided into active targeting, passive targeting, and physical and chemical targeting. In terms of liver targeting, because of specificity, active liver-targeting liposomes have received more and more attention, and these types of liposomes can be used in liver fibrosis, hepatitis and other chronic liver diseases. In addition, the particle size could control the passive liver targeting of liposomes, while the liver-targeted liposomes of the physical and chemical targeting type have advantages in treating hepatic carcinoma. In this paper, we focus on the basics and application of liver-targeting liposome drug delivery system in hepatic diseases.
Collapse
|
18
|
Xu J, Chen Y, Deng L, Liu J, Cao Y, Li P, Ran H, Zheng Y, Wang Z. Microwave-activated nanodroplet vaporization for highly efficient tumor ablation with real-time monitoring performance. Biomaterials 2016; 106:264-75. [PMID: 27573134 DOI: 10.1016/j.biomaterials.2016.08.034] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Revised: 08/16/2016] [Accepted: 08/18/2016] [Indexed: 12/31/2022]
|
19
|
Jiang R, Meng L, Sun L, He X, Liang X, Zhang J, Zhang Z. Microwave ablation-assisted liver gene transfection in rats. Int J Hyperthermia 2016; 32:666-72. [PMID: 27362775 DOI: 10.1080/02656736.2016.1195019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Affiliation(s)
- Ruoyu Jiang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
| | - Lingkai Meng
- Department of General Surgery, Tianjin Medical University Hospital for Metabolic Diseases, Tianjin Medical University, Tianjin, China
| | - Longhao Sun
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
| | - Xianghui He
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
| | - Xiaoyu Liang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
| | - Jie Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
| | - Zhixiang Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
| |
Collapse
|
20
|
Wang S, Mei XG, Goldberg SN, Ahmed M, Lee JC, Gong W, Han HB, Yan K, Yang W. Does Thermosensitive Liposomal Vinorelbine Improve End-Point Survival after Percutaneous Radiofrequency Ablation of Liver Tumors in a Mouse Model? Radiology 2016; 279:762-72. [PMID: 26785043 DOI: 10.1148/radiol.2015150787] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Purpose To investigate the role of thermosensitive liposome-encapsulated vinorelbine (Thermo-Vin) in combined radiofrequency (RF) ablation of liver tumors. Materials and Methods Approval from the institutional animal care and use committee was obtained before this study. First, the anticancer efficacy of Thermo-Vin was assessed in vitro (H22 cells) for 72 hours at 37°C or 42°C. Next, 203 H22 liver adenocarcinomas were implanted in 191 mice for in vivo study. Tumors were randomized into seven groups: (a) no treatment, (b) treatment with RF ablation alone, (c) treatment with RF ablation followed by free vinorelbine (Free-Vin) at 30 minutes, (d) treatment with RF ablation followed by empty liposomes (Empty-Lip+RF), (e) treatment with RF ablation followed by Thermo-Vin (5 mg/kg), (f) treatment with RF ablation followed by Thermo-Vin (10 mg/kg), and (g) treatment with RF ablation followed by Thermo-Vin (20 mg/kg). Tumor destruction areas and pathologic changes were compared for different groups at 24 and 72 hours after treatment. Kaplan-Meier analysis was used to compare end-point survival (tumor < 30 mm in diameter). Additionally, the effect of initial tumor size on long-term outcome was analyzed. Results In vitro, both Free-Vin and Thermo-Vin dramatically inhibited H22 cell viability at 24 hours. Likewise, in vivo, 10 mg/kg Thermo-Vin+RF ablation increased tumor destruction compared with RF ablation (P = .001). Intratumoral vinorelbine accumulation with Thermo-Vin+RF increased 15-fold compared with Free-Vin alone. Thermo-Vin substantially increased apoptosis at the coagulation margin and suppressed cellular proliferation in the residual tumor (P < .001). The Thermo-Vin+RF study arm also had better survival than the arm treated with RF ablation alone (mean, 37.6 days ± 20.1 vs 23.4 days ± 5.0; P = .001), the arm treated with Free-Vin+RF (23.3 days ± 1.2, P = .002), or the arm treated with Empty-Lip+RF (20.8 days ± 0.4, P < .001) in animals with medium-sized (10-12-mm) tumors. No significant difference in end-point survival was noted in the treatment arms with large or small tumors. Conclusion Thermo-Vin can effectively increase tumor destruction and improve animal survival. End-point survival is most affected in animals with medium-sized tumors, suggesting that combination therapy should be tailored to tumor size and the expected volume of ablation of the device used. (©) RSNA, 2016 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Song Wang
- From the Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Ultrasound (S.W., J.C.L., K.Y., W.Y.) and Department of Biobank (H.B.H.), Peking University Cancer Hospital and Institute, 52 Fucheng Rd, Haidian District, Beijing 100142, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China (X.G.M., W.G.); Division of Image-guided Therapy, Department of Radiology, Hadassah Hebrew University Medical Center, Jerusalem, Israel (S.N.G.); and Laboratory for Minimally Invasive Tumor Therapies, Department of Radiology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, Mass (S.N.G., M.A.)
| | - Xing-Guo Mei
- From the Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Ultrasound (S.W., J.C.L., K.Y., W.Y.) and Department of Biobank (H.B.H.), Peking University Cancer Hospital and Institute, 52 Fucheng Rd, Haidian District, Beijing 100142, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China (X.G.M., W.G.); Division of Image-guided Therapy, Department of Radiology, Hadassah Hebrew University Medical Center, Jerusalem, Israel (S.N.G.); and Laboratory for Minimally Invasive Tumor Therapies, Department of Radiology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, Mass (S.N.G., M.A.)
| | - S Nahum Goldberg
- From the Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Ultrasound (S.W., J.C.L., K.Y., W.Y.) and Department of Biobank (H.B.H.), Peking University Cancer Hospital and Institute, 52 Fucheng Rd, Haidian District, Beijing 100142, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China (X.G.M., W.G.); Division of Image-guided Therapy, Department of Radiology, Hadassah Hebrew University Medical Center, Jerusalem, Israel (S.N.G.); and Laboratory for Minimally Invasive Tumor Therapies, Department of Radiology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, Mass (S.N.G., M.A.)
| | - Muneeb Ahmed
- From the Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Ultrasound (S.W., J.C.L., K.Y., W.Y.) and Department of Biobank (H.B.H.), Peking University Cancer Hospital and Institute, 52 Fucheng Rd, Haidian District, Beijing 100142, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China (X.G.M., W.G.); Division of Image-guided Therapy, Department of Radiology, Hadassah Hebrew University Medical Center, Jerusalem, Israel (S.N.G.); and Laboratory for Minimally Invasive Tumor Therapies, Department of Radiology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, Mass (S.N.G., M.A.)
| | - Jung-Chieh Lee
- From the Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Ultrasound (S.W., J.C.L., K.Y., W.Y.) and Department of Biobank (H.B.H.), Peking University Cancer Hospital and Institute, 52 Fucheng Rd, Haidian District, Beijing 100142, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China (X.G.M., W.G.); Division of Image-guided Therapy, Department of Radiology, Hadassah Hebrew University Medical Center, Jerusalem, Israel (S.N.G.); and Laboratory for Minimally Invasive Tumor Therapies, Department of Radiology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, Mass (S.N.G., M.A.)
| | - Wei Gong
- From the Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Ultrasound (S.W., J.C.L., K.Y., W.Y.) and Department of Biobank (H.B.H.), Peking University Cancer Hospital and Institute, 52 Fucheng Rd, Haidian District, Beijing 100142, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China (X.G.M., W.G.); Division of Image-guided Therapy, Department of Radiology, Hadassah Hebrew University Medical Center, Jerusalem, Israel (S.N.G.); and Laboratory for Minimally Invasive Tumor Therapies, Department of Radiology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, Mass (S.N.G., M.A.)
| | - Hai-Bo Han
- From the Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Ultrasound (S.W., J.C.L., K.Y., W.Y.) and Department of Biobank (H.B.H.), Peking University Cancer Hospital and Institute, 52 Fucheng Rd, Haidian District, Beijing 100142, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China (X.G.M., W.G.); Division of Image-guided Therapy, Department of Radiology, Hadassah Hebrew University Medical Center, Jerusalem, Israel (S.N.G.); and Laboratory for Minimally Invasive Tumor Therapies, Department of Radiology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, Mass (S.N.G., M.A.)
| | - Kun Yan
- From the Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Ultrasound (S.W., J.C.L., K.Y., W.Y.) and Department of Biobank (H.B.H.), Peking University Cancer Hospital and Institute, 52 Fucheng Rd, Haidian District, Beijing 100142, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China (X.G.M., W.G.); Division of Image-guided Therapy, Department of Radiology, Hadassah Hebrew University Medical Center, Jerusalem, Israel (S.N.G.); and Laboratory for Minimally Invasive Tumor Therapies, Department of Radiology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, Mass (S.N.G., M.A.)
| | - Wei Yang
- From the Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Ultrasound (S.W., J.C.L., K.Y., W.Y.) and Department of Biobank (H.B.H.), Peking University Cancer Hospital and Institute, 52 Fucheng Rd, Haidian District, Beijing 100142, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China (X.G.M., W.G.); Division of Image-guided Therapy, Department of Radiology, Hadassah Hebrew University Medical Center, Jerusalem, Israel (S.N.G.); and Laboratory for Minimally Invasive Tumor Therapies, Department of Radiology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, Mass (S.N.G., M.A.)
| |
Collapse
|
21
|
Dicheva BM, ten Hagen TLM, Seynhaeve ALB, Amin M, Eggermont AMM, Koning GA. Enhanced Specificity and Drug Delivery in Tumors by cRGD-Anchoring Thermosensitive Liposomes. Pharm Res 2015. [PMID: 26202516 PMCID: PMC4628091 DOI: 10.1007/s11095-015-1746-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Purpose To develop RGD-targeted thermosensitive liposomes with increased tumor retention, improving drug release efficiency upon mild hyperthermia (HT) in both tumor and angiogenic endothelial cells. Methods Standard termosensitive liposomes (TSL) and TSL containing a cyclic Arg-Gly-Asp (cRGD) pentapeptide with the sequence Arg-Cys-D-Phe-Asp-Gly (RGDf[N-Met]C) were synthetized, loaded with Dox and characterized. Temperature- and time-dependent drug release profiles were assessed by fluorometry. Intracellular Dox delivery was studied by flow cytometry and confocal microscopy. Cytotoxic effect of TSL and RGD-TSL was studied on B16Bl6 melanoma, B16F10 melanoma and HUVEC. Intravital microscopy was performed on B16Bl6 tumors implanted in dorsal-skin fold window-bearing mice. Pharmacokinetic and biodistribution of Dox-TSL and Dox-RGD-TSL were followed in B16Bl6 tumor bearing mice upon normothermia or initial hyperthermia conditions. Results DLS and cryo-TEM revealed particle homogeneity and size of around 85 nm. Doxorubicin loading efficiency was >95%as assessed by spectrofluorometry. Flow cytometry and confocal microscopy showed a specific uptake of RGD-TSL by melanoma and endothelial cells when compared to TSL and an increased doxorubicin delivery. High resolution intravital microscopy demonstrated specific accumulation of RGD-TSL to the tumor vasculature. Moreover, application of hyperthermia resulted in massive drug release from RGD-TSL. Biodistribution studies showed that initial hyperthermia increases Dox uptake in tumors from TSL and RGD-TSL. Conclusion RGD-TSL have potency to increase drug efficacy due to higher uptake by tumor and angiogenic endothelial cells in combination with heat-triggered drug release. Electronic supplementary material The online version of this article (doi:10.1007/s11095-015-1746-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bilyana M Dicheva
- Laboratory Experimental Surgical Oncology, Section Surgical Oncology Department of Surgery, Erasmus MC Cancer Center, Rotterdam, The Netherlands. .,Laboratory of Experimental Surgical Oncology, Department of Surgical Oncology, Erasmus MC Cancer Institute, PO Box 2040, 3000, CA, Rotterdam, The Netherlands.
| | - Timo L M ten Hagen
- Laboratory Experimental Surgical Oncology, Section Surgical Oncology Department of Surgery, Erasmus MC Cancer Center, Rotterdam, The Netherlands
| | - Ann L B Seynhaeve
- Laboratory Experimental Surgical Oncology, Section Surgical Oncology Department of Surgery, Erasmus MC Cancer Center, Rotterdam, The Netherlands
| | - Mohamadreza Amin
- Laboratory Experimental Surgical Oncology, Section Surgical Oncology Department of Surgery, Erasmus MC Cancer Center, Rotterdam, The Netherlands.,Department of Pharmaceutics, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alexander M M Eggermont
- Laboratory Experimental Surgical Oncology, Section Surgical Oncology Department of Surgery, Erasmus MC Cancer Center, Rotterdam, The Netherlands.,Institut Gustave Roussy, Villejuif, France
| | - Gerben A Koning
- Laboratory Experimental Surgical Oncology, Section Surgical Oncology Department of Surgery, Erasmus MC Cancer Center, Rotterdam, The Netherlands
| |
Collapse
|
22
|
Evans SS, Repasky EA, Fisher DT. Fever and the thermal regulation of immunity: the immune system feels the heat. Nat Rev Immunol 2015; 15:335-49. [PMID: 25976513 PMCID: PMC4786079 DOI: 10.1038/nri3843] [Citation(s) in RCA: 678] [Impact Index Per Article: 67.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Fever is a cardinal response to infection that has been conserved in warm-blooded and cold-blooded vertebrates for more than 600 million years of evolution. The fever response is executed by integrated physiological and neuronal circuitry and confers a survival benefit during infection. In this Review, we discuss our current understanding of how the inflammatory cues delivered by the thermal element of fever stimulate innate and adaptive immune responses. We further highlight the unexpected multiplicity of roles of the pyrogenic cytokine interleukin-6 (IL-6), both during fever induction and during the mobilization of lymphocytes to the lymphoid organs that are the staging ground for immune defence. We also discuss the emerging evidence suggesting that the adrenergic signalling pathways associated with thermogenesis shape immune cell function.
Collapse
Affiliation(s)
- Sharon S Evans
- Department of Immunology, Roswell Park Cancer Institute, Elm &Carlton Streets, Buffalo, New York 14263, USA
| | - Elizabeth A Repasky
- Department of Immunology, Roswell Park Cancer Institute, Elm &Carlton Streets, Buffalo, New York 14263, USA
| | - Daniel T Fisher
- Department of Immunology, Roswell Park Cancer Institute, Elm &Carlton Streets, Buffalo, New York 14263, USA
| |
Collapse
|
23
|
Forbes N, Shin JE, Ogunyankin M, Zasadzinski JA. Inside-outside self-assembly of light-activated fast-release liposomes. Phys Chem Chem Phys 2015; 17:15569-78. [PMID: 25729792 DOI: 10.1039/c4cp05881j] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Building additional functionality into both the membrane and the internal compartments of biocompatible liposomes by self-assembly can provide ways of enhancing colloidal stability and spatial and temporal control of contents release. An interdigitation-fusion process is used to encapsulate near infrared light absorbing copper sulfide nanoparticles in the interior compartments of dipalmitoylphosphatidylcholine and dipalmitoylphosphatidylglycerol liposomes. Once formed, the liposome membrane is modified to include lysolipids and polyethylene glycol lipids by partitioning from lysolipid and PEG-lipid micelles in solution. This results in sterically stable, thermosensitive liposomes with a permeability transition near physiological temperature that can be triggered by NIR light irradiation. Rapid changes in local concentration can be induced with spatial and temporal control using NIR laser light.
Collapse
Affiliation(s)
- Natalie Forbes
- Department of Chemical Engineering, University of California, Santa Barbara, CA 93106, USA
| | | | | | | |
Collapse
|
24
|
Rizzitelli S, Giustetto P, Cutrin JC, Delli Castelli D, Boffa C, Ruzza M, Menchise V, Molinari F, Aime S, Terreno E. Sonosensitive theranostic liposomes for preclinical in vivo MRI-guided visualization of doxorubicin release stimulated by pulsed low intensity non-focused ultrasound. J Control Release 2015; 202:21-30. [PMID: 25626083 DOI: 10.1016/j.jconrel.2015.01.028] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 01/22/2015] [Accepted: 01/23/2015] [Indexed: 11/24/2022]
Abstract
The main goal of this study was to assess the theranostic performance of a nanomedicine able to generate MRI contrast as a response to the release from liposomes of the antitumor drug Doxorubicin triggered by the local exposure to pulsed low intensity non focused ultrasounds (pLINFU). In vitro experiments showed that Gadoteridol was an excellent imaging agent for probing the release of Doxorubicin following pLINFU stimulation. On this basis, the theranostic system was investigated in vivo on a syngeneic murine model of TS/A breast cancer. MRI offered an excellent guidance for monitoring the pLINFU-stimulated release of the drug. Moreover, it provided: i) an in vivo proof of the effective release of the liposomal content, and ii) a confirmation of the therapeutic benefits of the overall protocol. Ex vivo fluorescence microscopy indicated that the good therapeutic outcome was originated from a better diffusion of the drug in the tumor following the pLINFU stimulus. Very interestingly, the broad diffusion of the drug in the tumor stroma appeared to be mediated by the presence of the liposomes themselves. The results of this study highlighted either the great potential of US-based stimuli to safely trigger the release of a drug from its nanocarrier or the associated significant therapeutic improvement. Finally, MRI demonstrated to be a valuable technique to support chemotherapy and monitoring the outcome. Furthermore, in this specific case, the theranostic agent developed has a high clinical translatability because the MRI agent utilized is already approved for human use.
Collapse
Affiliation(s)
- S Rizzitelli
- Center for Molecular Imaging, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy
| | - P Giustetto
- Center for Molecular Imaging, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; Center for Preclinical Imaging, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Ribes 5, 10010 Colleretto Giacosa (TO), Italy
| | - J C Cutrin
- Center for Molecular Imaging, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy
| | - D Delli Castelli
- Center for Molecular Imaging, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy
| | - C Boffa
- Center for Molecular Imaging, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy
| | - M Ruzza
- Center for Molecular Imaging, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy
| | - V Menchise
- Institute for Biostructures and Bioimages (CNR) c/o Molecular Biotechnology Center, University of Torino, Italy
| | - F Molinari
- Biolab, Department of Electronics and Telecommunications, Politecnico di Torino, Torino, Italy
| | - S Aime
- Center for Molecular Imaging, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; Center for Preclinical Imaging, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Ribes 5, 10010 Colleretto Giacosa (TO), Italy
| | - E Terreno
- Center for Molecular Imaging, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; Center for Preclinical Imaging, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Ribes 5, 10010 Colleretto Giacosa (TO), Italy.
| |
Collapse
|
25
|
Dicheva BM, ten Hagen TL, Schipper D, Seynhaeve AL, van Rhoon GC, Eggermont AM, Koning GA. Targeted and heat-triggered doxorubicin delivery to tumors by dual targeted cationic thermosensitive liposomes. J Control Release 2014; 195:37-48. [DOI: 10.1016/j.jconrel.2014.07.058] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 07/22/2014] [Accepted: 07/30/2014] [Indexed: 11/28/2022]
|
26
|
Forbes N, Pallaoro A, Reich NO, Zasadzinski JA. Rapid, Reversible Release from Thermosensitive Liposomes Triggered by Near-Infra-Red Light. PARTICLE & PARTICLE SYSTEMS CHARACTERIZATION : MEASUREMENT AND DESCRIPTION OF PARTICLE PROPERTIES AND BEHAVIOR IN POWDERS AND OTHER DISPERSE SYSTEMS 2014; 31:1158-1167. [PMID: 29167602 PMCID: PMC5695930 DOI: 10.1002/ppsc.201400035] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Indexed: 05/24/2023]
Abstract
We present a novel drug carrier consisting of plasmonic hollow gold nanoshells (HGN) chemically tethered to liposomes made temperature sensitive with lysolipids (LTSL). Continuous-wave irradiation by physiologically friendly near infra-red light at 800 nm for 2.5 minutes at laser intensities an order of magnitude below that known to damage skin generates heating localized to the liposome membrane. The heating increases the liposome permeability in an irradiation dose-dependent, but reversible manner, resulting in rapid release of small molecules such as the self-quenching dye carboxyfluorescein or the chemotherapeutic doxorubicin, without raising the bulk temperature. The local rise in nanoshell temperature under laser irradiation was inferred by comparing dye release rates from the LTSL via bulk heating to that induced by irradiation. Laser-irradiation of LTSL enables precise control of contents release with low temperature gradients confined to areas irradiated by the laser focus. The combined effects of rapid local release and localized hyperthermia provide a synergistic effect as shown by a near doubling of androgen resistant PPC-1 prostate cancer cell toxicity compared to the same concentration of free doxorubicin.
Collapse
Affiliation(s)
- Natalie Forbes
- Department of Chemical Engineering, University of California, Santa Barbara, CA 93106
| | - Alessia Pallaoro
- Department of Chemistry, University of California, Santa Barbara, CA 93106
| | - Norbert O Reich
- Department of Chemistry, University of California, Santa Barbara, CA 93106
| | - Joseph A Zasadzinski
- Chemical Engineering and Material Science, University of Minnesota, Minneapolis, MN 55455
| |
Collapse
|
27
|
Shih YH, Lin XZ, Yeh CH, Peng CL, Shieh MJ, Lin WJ, Luo TY. Preparation and therapeutic evaluation of (188)Re-thermogelling emulsion in rat model of hepatocellular carcinoma. Int J Nanomedicine 2014; 9:4191-201. [PMID: 25214783 PMCID: PMC4159399 DOI: 10.2147/ijn.s66346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Radiolabeled Lipiodol® (Guerbet, Villepinte, France) is routinely used in hepatoma therapy. The temperature-sensitive hydrogel polyethylene glycol-b-poly-DL-lactic acid-co-glycolic acid-b-polyethylene glycol triblock copolymer is used as an embolic agent and sustained drug release system. This study attempted to combine the polyethylene glycol-b-poly-DL-lactic acid-co-glycolic acid-b-polyethylene glycol hydrogel and radio-labeled Lipiodol to form a new radio-thermogelling emulsion, rhenium-188–N,N’-1,2-ethanediylbis-L-cysteine diethyl-ester dihydrochloride–Lipiodol/hydrogel (188Re-ELH). The therapeutic potential of 188Re-ELH was evaluated in a rodent hepatoma model. Rhenium-188 chelated with N,N’-1,2-ethanediylbis-L-cysteine diethyl-ester dihydrochloride was extracted with Lipiodol to obtain rhenium-188–N,N’-1,2-ethanediylbis-L-cysteine diethyl-ester dihydrochloride–Lipiodol (188Re-EL), which was blended with the hydrogel in equal volumes to develop 188Re-ELH. The 188Re-ELH phase stability was evaluated at different temperatures. Biodistribution patterns and micro-single-photon emission computed tomography/computed tomography images in Sprague Dawley rats implanted with the rat hepatoma cell line N1-S1 were observed after in situ tumoral injection of ~3.7 MBq 188Re-ELH. The therapeutic potential of 188Re-EL (48.58±3.86 MBq/0.1 mL, n=12) was evaluated in a 2-month survival study using the same animal model. The therapeutic effects of 188Re-ELH (25.52±4.64 MBq/0.1 mL, n=12) were evaluated and compared with those of 188Re-EL. The responses were assessed by changes in tumor size and survival rates. The 188Re-ELH emulsion was stable in the gel form at 25°C–35°C for >52 hours. Biodistribution data and micro-single-photon emission computed tomography/computed tomography images of the 188Re-ELH group indicated that most activity was selectively observed in hepatomas. Long-term 188Re-ELH studies have demonstrated protracted reductions in tumor volumes and positive effects on the survival rates (75%) of N1-S1 hepatoma-bearing rats. Conversely, the 2-month survival rate was 13% in the control sham group. Therapeutic responses differed significantly between the two groups (P<0.005). Thus, the hydrogel enhanced the injection stability of 188Re-EL in an animal hepatoma model. Given the synergistic results, direct 188Re-ELH intratumoral injection is a potential therapeutic alternative for hepatoma treatment.
Collapse
Affiliation(s)
- Ying-Hsia Shih
- Isotope Application Division, Institute of Nuclear Energy Research, Longtan, Taiwan ; Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
| | - Xi-Zhang Lin
- Department of Internal Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chung-Hsin Yeh
- Isotope Application Division, Institute of Nuclear Energy Research, Longtan, Taiwan
| | - Cheng-Liang Peng
- Isotope Application Division, Institute of Nuclear Energy Research, Longtan, Taiwan ; Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
| | - Ming-Jium Shieh
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan ; Department of Oncology, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Wuu-Jyh Lin
- Isotope Application Division, Institute of Nuclear Energy Research, Longtan, Taiwan
| | - Tsai-Yueh Luo
- Isotope Application Division, Institute of Nuclear Energy Research, Longtan, Taiwan ; Institute of Radiological Science, Central Taiwan University of Science and Technology, Taichung, Taiwan
| |
Collapse
|
28
|
Zochowska M, Piguet AC, Jemielity J, Kowalska J, Szolajska E, Dufour JF, Chroboczek J. Virus-like particle-mediated intracellular delivery of mRNA cap analog with in vivo activity against hepatocellular carcinoma. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2014; 11:67-76. [PMID: 25101883 DOI: 10.1016/j.nano.2014.07.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 06/17/2014] [Accepted: 07/23/2014] [Indexed: 12/21/2022]
Abstract
Adenovirus dodecahedron (Dd), a nanoparticulate proteinaceous biodegradable virus-like particle (VLP), was used as a vector for delivery of an oncogene inhibitor to hepatocellular carcinoma (HCC) rat orthotopic model. Initiation factor eIF4E is an oncogene with elevated expression in human cancers. Cell-impermeant eIF4E inhibitor, cap structure analog (cap) and anti-cancer antibiotic doxorubicin (Dox) were delivered as Dd conjugates. Dd-cap and Dd-dox inhibited cancer cell culture proliferation up to 50 and 84%, respectively, while with free Dox similar results could be obtained only at a 5 times higher concentration. In animal HCC model the combination treatment of Dd-cap/Dd-dox caused 40% inhibition of tumor growth. Importantly, the level of two pro-oncogenes, eIF4E and c-myc, was significantly diminished in tumor sections of treated rats. Attachment to Dd, a virus-like particle, permitted the first demonstration of cap analog intracellular delivery and resulted in improved doxorubicin delivery leading to statistically significant inhibition of HCC tumor growth.
Collapse
Affiliation(s)
- Monika Zochowska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Anne-Christine Piguet
- Hepatology, Department of Clinical Research, University of Berne, Berne, Switzerland
| | - Jacek Jemielity
- Division of Biophysics, Institute of Experimental Physics, Faculty of Physics, University of Warsaw, Warsaw, Poland; Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | - Joanna Kowalska
- Division of Biophysics, Institute of Experimental Physics, Faculty of Physics, University of Warsaw, Warsaw, Poland
| | - Ewa Szolajska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Jean-François Dufour
- University Clinics of Visceral Surgery and Medicine, Inselspital Berne, Berne, Switzerland
| | - Jadwiga Chroboczek
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland; Therex, TIMC-IMAG, CNRS, Université Joseph Fourier, La Tronche, France.
| |
Collapse
|
29
|
Investigation on nanoparticle distribution for thermal ablation of a tumour subjected to nanoparticle assisted thermal therapy. J Therm Biol 2014; 43:70-80. [PMID: 24956960 DOI: 10.1016/j.jtherbio.2014.05.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Revised: 04/16/2014] [Accepted: 05/12/2014] [Indexed: 02/08/2023]
Abstract
This study investigates the effect of the distribution of nanoparticles delivered to a skin tumour for the thermal ablation conditions attained during thermal therapy. Ultimate aim is to define a distribution of nanoparticles as well as a combination of other therapeutic parameters to attain thermal ablation temperatures (50-60 °C) within whole of the tumour region. Three different cases of nanoparticle distributions are analysed under controlled conditions for all other parameters viz. irradiation intensity and duration, and volume fraction of nanoparticles. Results show that distribution of nanoparticles into only the periphery of tumour resulted in desired thermal ablation temperature in whole of tumour. For the tumour size considered in this study, an irradiation intensity of 1.25 W/cm(2) for duration of 300 s and a nanoparticle volume fraction of 0.001% was optimal to attain a temperature of ≥53 °C within the whole tumour region. It is concluded that distribution of nanoparticles in peripheral region of tumour, along with a controlled combination of other parameters, seems favourable and provides a promising pathway for thermal ablation of a tumour subjected to nanoparticle assisted thermal therapy.
Collapse
|